1
|
Khormizi FZ, Saadi HF, Khatami M, Heidari MM, Tabrizi F, Hashemi A, Khanjarpanah Z. Identification of rare and pathogenic TAL2 gene mutations in B-lineage acute lymphoblastic leukemia (B-ALL) using mutational screening and comprehensive bioinformatics analysis. Mol Biol Rep 2025; 52:125. [PMID: 39821746 DOI: 10.1007/s11033-025-10229-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
BACKGROUND Recent genomic research has identified several genetic factors contributing to B-cell acute lymphoblastic leukemia (B-ALL). However, the exact cause of the disease is still not fully understood. It is known that mutations in the TAL2 gene play important roles in the development of acute lymphoblastic leukemia. This study aimed to analyze the molecular and computational profile of the TAL2 mutations in a group of Iranian B-ALL patients for the first time. METHODS AND RESULTS In this study, 188 patients were enrolled, and the TAL2 gene was sequenced to identify gene variations. The study included structural/functional analysis, homology modeling, molecular docking, and molecular dynamics (MD) simulations to assess the potential impact of the missense mutations on the protein's structure. Three nucleotide variations in the exon, three variations in the 3'UTR, and one deletion variant in the 3'UTR were detected in patients. Through in-silico analysis, it was found that the p. Asp35Glu missense mutation is located in the bHLH domain of the TAL2 protein. Also, the structural and functional analyses predicted that this mutation is a pathogenic or likely pathogenic variant in B-ALL patients. Moreover, a multiple nucleotide deletion (g.659_668del) was found in the 3'UTR in most patients. This deletion occurs at the site of poly-A tail attachment and appears to have significant implications. CONCLUSIONS These findings offer new insights into the impact of genetic variants in the TAL2 gene on the development of B-ALL and their potential role as tumor biomarkers for the B-ALL. Further research is needed to explore the relationship between specific TAL2 mutations and the clinical presentation of B-ALL.
Collapse
Affiliation(s)
| | | | | | | | | | - Azam Hashemi
- Hematology and Oncology Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zohre Khanjarpanah
- Hematology and Oncology Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
2
|
Xu X, Chen J, Zhao H, Pi Y, Lin G, Hu L. Single-Cell RNA-seq Analysis of a Human Embryonic Stem Cell to Endothelial Cell System Based on Transcription Factor Overexpression. Stem Cell Rev Rep 2023; 19:2497-2509. [PMID: 37537495 DOI: 10.1007/s12015-023-10598-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 08/05/2023]
Abstract
BACKGROUND Human embryonic stem cell (hESC)-derived endothelial cells (ECs) possess therapeutic potential in many diseases. Cytokine supplementation induction and transcription factor overexpression have become two mainstream methods of hESC-EC induction. Single-cell RNA-seq technology has been widely used to analyse dynamic processes during hESC-EC induction and components of induced endothelial cells. However, studies that used single-cell RNA-seq are mainly based on cytokine supplementation methods. In this study, we used a high-efficiency human embryonic stem cell-endothelial cell line (hESC-EC) called the "FLI1-PKC system" as a research model and employed single-cell RNA sequencing (scRNA-seq) to investigate the transcriptional landscape and cellular dynamics. METHODS The high-efficiency hESC-EC induction (FLI1-PKC) system was established in our previous study. We applied single-cell RNA sequencing (scRNA-seq) of the differentiated cells at different time points and investigated the gene expression profiles. RESULTS The FLI1-PKC induction system can directionally differentiate hESCs into mature endothelial cells with all the requisite functions. Unlike other hES-EC induction protocols, the FLI1-PKC method follows a different induction route; nonetheless, the transcriptome of induced endothelial cells (iECs) remains the same. The elevated number of activated transcription factors may explain why the FLI1-PKC system is more effective than other hES-EC protocols. CONCLUSION Our study has presented a single-cell transcriptional overview of a high-efficiency hESC-EC induction system, which can be used as a model and reference for further improvement in other hESC induction systems.
Collapse
Affiliation(s)
- XiangWang Xu
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, People's Republic of China
| | - JunRu Chen
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Hunan, 410008, Changsha, China
| | - Hao Zhao
- Xiangya Hospital of Central South University, Changsha, Human, China
| | - YuZe Pi
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, People's Republic of China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, People's Republic of China
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Hunan, 410008, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, National Engineering and Research Center of Human Stem Cell, Changsha, 410008, Hunan, China
| | - Liang Hu
- Institute of Reproductive and Stem Cell Engineering, NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Science, Central South University, Xiangya Road 88#, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Hunan, 410008, Changsha, China.
- Hunan International Scientific and Technological Cooperation Base of Development and Carcinogenesis, National Engineering and Research Center of Human Stem Cell, Changsha, 410008, Hunan, China.
| |
Collapse
|
3
|
Farries G, Bryan K, McGivney CL, McGettigan PA, Gough KF, Browne JA, MacHugh DE, Katz LM, Hill EW. Expression Quantitative Trait Loci in Equine Skeletal Muscle Reveals Heritable Variation in Metabolism and the Training Responsive Transcriptome. Front Genet 2019; 10:1215. [PMID: 31850069 PMCID: PMC6902038 DOI: 10.3389/fgene.2019.01215] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/04/2019] [Indexed: 01/10/2023] Open
Abstract
While over ten thousand genetic loci have been associated with phenotypic traits and inherited diseases in genome-wide association studies, in most cases only a relatively small proportion of the trait heritability is explained and biological mechanisms underpinning these traits have not been clearly identified. Expression quantitative trait loci (eQTL) are subsets of genomic loci shown experimentally to influence gene expression. Since gene expression is one of the primary determinants of phenotype, the identification of eQTL may reveal biologically relevant loci and provide functional links between genomic variants, gene expression and ultimately phenotype. Skeletal muscle (gluteus medius) gene expression was quantified by RNA-seq for 111 Thoroughbreds (47 male, 64 female) in race training at a single training establishment sampled at two time-points: at rest (n = 92) and four hours after high-intensity exercise (n = 77); n = 60 were sampled at both time points. Genotypes were generated from the Illumina Equine SNP70 BeadChip. Applying a False Discovery Rate (FDR) corrected P-value threshold (PFDR < 0.05), association tests identified 3,583 cis-eQTL associated with expression of 1,456 genes at rest; 4,992 cis-eQTL associated with the expression of 1,922 genes post-exercise; 1,703 trans-eQTL associated with 563 genes at rest; and 1,219 trans-eQTL associated with 425 genes post-exercise. The gene with the highest cis-eQTL association at both time-points was the endosome-associated-trafficking regulator 1 gene (ENTR1; Rest: PFDR = 3.81 × 10-27, Post-exercise: PFDR = 1.66 × 10-24), which has a potential role in the transcriptional regulation of the solute carrier family 2 member 1 glucose transporter protein (SLC2A1). Functional analysis of genes with significant eQTL revealed significant enrichment for cofactor metabolic processes. These results suggest heritable variation in genomic elements such as regulatory sequences (e.g. gene promoters, enhancers, silencers), microRNA and transcription factor genes, which are associated with metabolic function and may have roles in determining end-point muscle and athletic performance phenotypes in Thoroughbred horses. The incorporation of the eQTL identified with genome and transcriptome-wide association may reveal useful biological links between genetic variants and their impact on traits of interest, such as elite racing performance and adaptation to training.
Collapse
Affiliation(s)
- Gabriella Farries
- UCD School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Kenneth Bryan
- UCD School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | | | - Paul A McGettigan
- UCD School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Katie F Gough
- UCD School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - John A Browne
- UCD School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - David E MacHugh
- UCD School of Agriculture and Food Science, University College Dublin, Dublin, Ireland.,UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Lisa Michelle Katz
- UCD School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Emmeline W Hill
- UCD School of Agriculture and Food Science, University College Dublin, Dublin, Ireland.,Research and Development, Plusvital Ltd., Dublin, Ireland
| |
Collapse
|
4
|
Cang J, Savier E, Barchini J, Liu X. Visual Function, Organization, and Development of the Mouse Superior Colliculus. Annu Rev Vis Sci 2018; 4:239-262. [PMID: 29852095 DOI: 10.1146/annurev-vision-091517-034142] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The superior colliculus (SC) is the most prominent visual center in mice. Studies over the past decade have greatly advanced our understanding of the function, organization, and development of the mouse SC, which has rapidly become a popular model in vision research. These studies have described the diverse and cell-type-specific visual response properties in the mouse SC, revealed their laminar and topographic organizations, and linked the mouse SC and downstream pathways with visually guided behaviors. Here, we summarize these findings, compare them with the rich literature of SC studies in other species, and highlight important gaps and exciting future directions. Given its clear importance in mouse vision and the available modern neuroscience tools, the mouse SC holds great promise for understanding the cellular, circuit, and developmental mechanisms that underlie visual processing, sensorimotor transformation, and, ultimately, behavior.
Collapse
Affiliation(s)
- Jianhua Cang
- Department of Biology and Department of Psychology, University of Virginia, Charlottesville, Virginia 22904, USA;
| | - Elise Savier
- Department of Biology and Department of Psychology, University of Virginia, Charlottesville, Virginia 22904, USA;
| | - Jad Barchini
- Department of Functional Architecture and Development of Cerebral Cortex, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458, USA
| | - Xiaorong Liu
- Department of Biology and Department of Psychology, University of Virginia, Charlottesville, Virginia 22904, USA;
| |
Collapse
|
5
|
Kobayashi T. Expression and Regulation of Tal2 during Neuronal Differentiation in P19 Cells. YAKUGAKU ZASSHI 2017; 137:61-71. [PMID: 28049897 DOI: 10.1248/yakushi.16-00176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
T-cell acute lymphocytic leukemia 2 (Tal2) is a gene encoding a member of the basic helix-loop-helix transcription factor family, which is essential for the normal development of the mouse brain. We found that Tal2 was induced during neural differentiation in P19 cells, which are pluripotent mouse embryonal carcinoma cells that differentiate into the neural lineage upon both exposure to all-trans retinoic acid (atRA) and the formation of cell aggregation. Tal2 expression during neural differentiation in P19 cells was detected within 3 h after induction with atRA and retinoic acid receptor α (RARα). The atRA-RARα complex is known to bind to a characteristic retinoic acid response element (RARE) located in the promoter of target genes. We found a RARE-like element in the intron of Tal2. We also found a TATA-box-like element in the 5' region. The TATA-box-like element functioned as a core promoter, and TATA- box binding protein bound to this element upstream of Tal2 in P19 cells. The RARE-like element responded to atRA signaling that activated the transcription, and RARα was bound to this element in the intron of Tal2 in P19 cells. Furthermore, the interaction between these elements on Tal2 was confirmed in a chromatin immunoprecipitation assay. Because the neural differentiation of P19 cells mimics in part the development of the nervous system, P19 cells are useful for studying the mechanism underlying the role of Tal2 in neural differentiation. Further work is underway to clarify the function of Tal2 in neural differentiation using the differentiation system of P19 cells.
Collapse
|
6
|
High Energy Particle Radiation-associated Oncogenic Transformation in Normal Mice: Insight into the Connection between Activation of Oncotargets and Oncogene Addiction. Sci Rep 2016; 6:37623. [PMID: 27876887 PMCID: PMC5120307 DOI: 10.1038/srep37623] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 11/01/2016] [Indexed: 12/19/2022] Open
Abstract
Concerns on high-energy particle radiation-induced tumorigenic transformation of normal tissue in astronauts, and in cancer patients undergoing radiotherapy, emphasizes the significance of elucidating the mechanisms involved in radiogenic transformation processes. Mostly used genetically modified or tumor-prone models are less reliable in determining human health risk in space or protracted post-treatment normal tissue toxicity. Here, in wild type C57BL/6 mice, we related the deregulation of distinctive set of tissue-specific oncotargets in major organs upon 56Fe (600 MeV/amu; 0.5 Gy/min; 0.8 Gy) particle radiation and compared the response with low LET γ-radiation (137Cs; 0.5 Gy/min; 2 Gy). One of the novel findings is the ‘tissue-independent’ activation of TAL2 upon high-energy radiation, and thus qualifies TAL2 as a potential biomarker for particle and other qualities of radiation. Heightened expression of TAL2 gene transcript, which sustained over four weeks post-irradiation foster the concept of oncogene addiction signaling in radiogenic transformation. The positive/negative expression of other selected oncotargets that expresses tissue-dependent manner indicated their role as a secondary driving force that addresses the diversity of tissue-dependent characteristics of tumorigenesis. This study, while reporting novel findings on radiogenic transformation of normal tissue when exposed to particle radiation, it also provides a platform for further investigation into different radiation quality, LET and dose/dose rate effect in healthy organs.
Collapse
|
7
|
Durning SP, Flanagan-Steet H, Prasad N, Wells L. O-Linked β-N-acetylglucosamine (O-GlcNAc) Acts as a Glucose Sensor to Epigenetically Regulate the Insulin Gene in Pancreatic Beta Cells. J Biol Chem 2015; 291:2107-18. [PMID: 26598517 DOI: 10.1074/jbc.m115.693580] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Indexed: 11/06/2022] Open
Abstract
The post-translational protein modification O-linked β-N-acetylglucosamine (O-GlcNAc) is a proposed nutrient sensor that has been shown to regulate multiple biological pathways. This dynamic and inducible enzymatic modification to intracellular proteins utilizes the end product of the nutrient sensing hexosamine biosynthetic pathway, UDP-GlcNAc, as its substrate donor. Type II diabetic patients have elevated O-GlcNAc-modified proteins within pancreatic beta cells due to chronic hyperglycemia-induced glucose overload, but a molecular role for O-GlcNAc within beta cells remains unclear. Using directed pharmacological approaches in the mouse insulinoma-6 (Min6) cell line, we demonstrate that elevating nuclear O-GlcNAc increases intracellular insulin levels and preserves glucose-stimulated insulin secretion during chronic hyperglycemia. The molecular mechanism for these observed changes appears to be, at least in part, due to elevated O-GlcNAc-dependent increases in Ins1 and Ins2 mRNA levels via elevations in histone H3 transcriptional activation marks. Furthermore, RNA deep sequencing reveals that this mechanism of altered gene transcription is restricted and that the majority of genes regulated by elevated O-GlcNAc levels are similarly regulated by a shift from euglycemic to hyperglycemic conditions. These findings implicate the O-GlcNAc modification as a potential mechanism for hyperglycemic-regulated gene expression in the beta cell.
Collapse
Affiliation(s)
- Sean P Durning
- From the Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602-1516 and
| | - Heather Flanagan-Steet
- From the Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602-1516 and
| | - Nripesh Prasad
- HudsonAlpha Institute of Biotechnology, Genomic Services Laboratory, Huntsville, Alabama 35806
| | - Lance Wells
- From the Complex Carbohydrate Research Center, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia 30602-1516 and
| |
Collapse
|
8
|
Kobayashi T, Suzuki M, Morikawa M, Kino K, Tanuma SI, Miyazawa H. Transcriptional regulation of Tal2 gene by all-trans retinoic acid (atRA) in P19 cells. Biol Pharm Bull 2014; 38:248-56. [PMID: 25482166 DOI: 10.1248/bpb.b14-00617] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
TAL2 is a transcription factor required in the normal development of mouse brain. In a previous study, we demonstrated that the expression of Tal2 gene is induced by the complex of all-trans retinoic acid (atRA) and retinoic acid receptor α (RARα) in mouse embryonal carcinoma P19 cells. atRA is also known to be important in inducing P19 cells to differentiate into the neural lineage. Therefore, we believe that the function of TAL2 in neural differentiation may be clarified by utilizing P19 cells. As the atRA-RARα complex induced the expression of Tal2, we focused on the regulatory region that is involved in its transcription. The atRA-RARα complex occupies a characteristic retinoic acid response element (RARE) located in the promoter of target genes. Therefore, we searched for RARE on the mouse Tal2 and found that a RARE-like element was located in the intron. We also found that a TATA-box-like element was located in the 5'-region of Tal2. Involvement between transcriptional activity and the TATA-box-like element was confirmed in the luciferase assay, and TATA-box binding protein was bound to this element upstream of Tal2 in P19 cells. atRA signaling activated the transcription through the RARE-like element, and RARα was bound to this element on Tal2 in P19 cells. In addition, the interaction between these elements on Tal2 was shown in the chromatin immunoprecipitation assay. These results suggest that the transcription of Tal2 is coordinately mediated by two distal regulatory elements.
Collapse
|
9
|
Kobayashi T, Komori R, Ishida K, Kino K, Tanuma SI, Miyazawa H. Tal2 expression is induced by all-trans retinoic acid in P19 cells prior to acquisition of neural fate. Sci Rep 2014; 4:4935. [PMID: 24816818 PMCID: PMC4017210 DOI: 10.1038/srep04935] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 04/24/2014] [Indexed: 11/10/2022] Open
Abstract
TAL2 is a member of the basic helix-loop-helix family and is essential for the normal development of the mouse brain. However, the function of TAL2 during brain development is unclear. P19 cells are pluripotent mouse embryonal carcinoma cells that adopt neural fates upon exposure to all-trans retinoic acid (atRA) and culture in suspension. We found that the expression of Tal2 gene was induced in P19 cells after addition of atRA in suspension culture. Tal2 expression was detected within 3 h after the induction, and had nearly returned to basal levels by 24 h. When GFP-tagged TAL2 (GFP-TAL2) was expressed in P19 cells, we observed GFP-TAL2 in the nucleus. Moreover, we showed that atRA and retinoic acid receptor α regulated Tal2 expression. These results demonstrate for the first time that atRA induces Tal2 expression in P19 cells, and suggest that TAL2 commits to the acquisition of neural fate in brain development.
Collapse
Affiliation(s)
- Takanobu Kobayashi
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki, Kagawa 769-2193, Japan
| | - Rie Komori
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki, Kagawa 769-2193, Japan
| | - Kiyoshi Ishida
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki, Kagawa 769-2193, Japan
| | - Katsuhito Kino
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki, Kagawa 769-2193, Japan
| | - Sei-ichi Tanuma
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Hiroshi Miyazawa
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, 1314-1 Shido, Sanuki, Kagawa 769-2193, Japan
| |
Collapse
|
10
|
Courtial N, Mücke C, Herkt S, Kolodziej S, Hussong H, Lausen J. The T-cell oncogene Tal2 Is a Target of PU.1 and upregulated during osteoclastogenesis. PLoS One 2013; 8:e76637. [PMID: 24086757 PMCID: PMC3784441 DOI: 10.1371/journal.pone.0076637] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 08/30/2013] [Indexed: 11/19/2022] Open
Abstract
Transcription factors play a crucial role in regulating differentiation processes during human life and are important in disease. The basic helix-loop-helix transcription factors Tal1 and Lyl1 play a major role in the regulation of gene expression in the hematopoietic system and are involved in human leukemia. Tal2, which belongs to the same family of transcription factors as Tal1 and Lyl1, is also involved in human leukaemia. However, little is known regarding the expression and regulation of Tal2 in hematopoietic cells. Here we show that Tal2 is expressed in hematopoietic cells of the myeloid lineage. Interestingly, we found that usage of the Tal2 promoter is different in human and mouse cells. Two promoters, hP1 and hP2 drive Tal2 expression in human erythroleukemia K562 cells, however in mouse RAW cells only the mP1 promoter is used. Furthermore, we found that Tal2 expression is upregulated during oesteoclastogenesis. We show that Tal2 is a direct target gene of the myeloid transcription factor PU.1, which is a key transcription factor for osteoclast gene expression. Strikingly, PU.1 binding to the P1 promoter is conserved between mouse and human, but PU.1 binding to P2 was only detected in human K562 cells. Additionally, we provide evidence that Tal2 influences the expression of the osteoclastic differentiation gene TRACP. These findings provide novel insight into the expression control of Tal2 in hematopoietic cells and reveal a function of Tal2 as a regulator of gene expression during osteoclast differentiation.
Collapse
Affiliation(s)
- Nadine Courtial
- Georg-Speyer-Haus, Institute for Biomedical Research, Frankfurt, Germany
| | - Christian Mücke
- Georg-Speyer-Haus, Institute for Biomedical Research, Frankfurt, Germany
| | - Stefanie Herkt
- Georg-Speyer-Haus, Institute for Biomedical Research, Frankfurt, Germany
| | - Stephan Kolodziej
- Georg-Speyer-Haus, Institute for Biomedical Research, Frankfurt, Germany
| | - Helge Hussong
- Georg-Speyer-Haus, Institute for Biomedical Research, Frankfurt, Germany
| | - Jörn Lausen
- Georg-Speyer-Haus, Institute for Biomedical Research, Frankfurt, Germany
- * E-mail:
| |
Collapse
|
11
|
Achim K, Peltopuro P, Lahti L, Tsai HH, Zachariah A, Astrand M, Salminen M, Rowitch D, Partanen J. The role of Tal2 and Tal1 in the differentiation of midbrain GABAergic neuron precursors. Biol Open 2013; 2:990-7. [PMID: 24167708 PMCID: PMC3798194 DOI: 10.1242/bio.20135041] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 06/26/2013] [Indexed: 12/22/2022] Open
Abstract
Midbrain- and hindbrain-derived GABAergic interneurons are critical for regulation of sleep, respiratory, sensory-motor and motivational processes, and they are implicated in human neurological disorders. However, the precise mechanisms that underlie generation of GABAergic neuron diversity in the midbrain–hindbrain region are poorly understood. Here, we show unique and overlapping requirements for the related bHLH proteins Tal1 and Tal2 in GABAergic neurogenesis in the midbrain. We show that Tal2 and Tal1 are specifically and sequentially activated during midbrain GABAergic neurogenesis. Similar to Gata2, a post-mitotic selector of the midbrain GABAergic neuron identity, Tal2 expression is activated very early during GABAergic neuron differentiation. Although the expression of Tal2 and Gata2 genes are independent of each other, Tal2 is important for normal midbrain GABAergic neurogenesis, possibly as a partner of Gata2. In the absence of Tal2, the majority of midbrain GABAergic neurons switch to a glutamatergic-like phenotype. In contrast, Tal1 expression is activated in a Gata2 and Tal2 dependent fashion in the more mature midbrain GABAergic neuron precursors, but Tal1 alone is not required for GABAergic neuron differentiation from the midbrain neuroepithelium. However, inactivation of both Tal2 and Tal1 in the developing midbrain suggests that the two factors co-operate to guide GABAergic neuron differentiation in a specific ventro-lateral midbrain domain. The observed similarities and differences between Tal1/Tal2 and Gata2 mutants suggest both co-operative and unique roles for these factors in determination of midbrain GABAergic neuron identities.
Collapse
Affiliation(s)
- Kaia Achim
- Department of Biosciences, P.O. Box 56, Viikinkaari 5, FIN00014-University of Helsinki , Helsinki , Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Ochocinska MJ, Muñoz EM, Veleri S, Weller JL, Coon SL, Pozdeyev N, Iuvone PM, Goebbels S, Furukawa T, Klein DC. NeuroD1 is required for survival of photoreceptors but not pinealocytes: results from targeted gene deletion studies. J Neurochem 2012; 123:44-59. [PMID: 22784109 DOI: 10.1111/j.1471-4159.2012.07870.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
NeuroD1 encodes a basic helix-loop-helix transcription factor involved in the development of neural and endocrine structures, including the retina and pineal gland. To determine the effect of NeuroD1 knockout in these tissues, a Cre/loxP recombination strategy was used to target a NeuroD1 floxed gene and generate NeuroD1 conditional knockout (cKO) mice. Tissue specificity was conferred using Cre recombinase expressed under the control of the promoter of Crx, which is selectively expressed in the pineal gland and retina. At 2 months of age, NeuroD1 cKO retinas have a dramatic reduction in rod- and cone-driven electroretinograms and contain shortened and disorganized outer segments; by 4 months, NeuroD1 cKO retinas are devoid of photoreceptors. In contrast, the NeuroD1 cKO pineal gland appears histologically normal. Microarray analysis of 2-month-old NeuroD1 cKO retina and pineal gland identified a subset of genes that were affected 2-100-fold; in addition, a small group of genes exhibit altered differential night/day expression. Included in the down-regulated genes are Aipl1, which is necessary to prevent retinal degeneration, and Ankrd33, whose protein product is selectively expressed in the outer segments. These findings suggest that NeuroD1 may act through Aipl1 and other genes to maintain photoreceptor homeostasis.
Collapse
Affiliation(s)
- Margaret J Ochocinska
- Section on Neuroendocrinology, Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Nakagawa Y, Shimogori T. Diversity of thalamic progenitor cells and postmitotic neurons. Eur J Neurosci 2012; 35:1554-62. [DOI: 10.1111/j.1460-9568.2012.08089.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
14
|
Suzuki-Hirano A, Ogawa M, Kataoka A, Yoshida AC, Itoh D, Ueno M, Blackshaw S, Shimogori T. Dynamic spatiotemporal gene expression in embryonic mouse thalamus. J Comp Neurol 2011; 519:528-43. [PMID: 21192082 DOI: 10.1002/cne.22531] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The anatomy of the mammalian thalamus is characterized by nuclei, which can be readily identified in postnatal animals. However, the molecular mechanisms that guide specification and differentiation of neurons in specific thalamic nuclei are still largely unknown, and few molecular markers are available for most of these thalamic subregions at early stages of development. We therefore searched for patterned gene expression restricted to specific mouse thalamic regions by in situ hybridization during the onset of thalamic neurogenesis (embryonic [E] days E10.5-E12.5). To obtain correct regional information, we used Shh as a landmark and compared spatial relationships with the zona limitans intrathalamica (Zli), the border of the p2 and p3 compartments of the diencephalon. We identified genes that are expressed specifically in the ventricular zone of the thalamic neuroepithelium and also identified a number of genes that already exhibited regional identity at E12.5. Although many genes expressed in the mantle regions of the thalamus at E12.5 showed regionally restricted patterns, none of these clearly corresponded to individual thalamic nuclei. We next examined gene expression at E15.5, when thalamocortical axons (TCAs) project from distinct regions of the thalamus and reach their targets in the cerebral cortex. Regionally restricted patterns of gene expression were again seen for many genes, but some regionally bounded expression patterns in the early postnatal thalamus had shifted substantially by E15.5. These findings reveal that nucleogenesis in the developing thalamus is associated with selective and complex changes in gene expression and provide a list of genes that may actively regulate the development of thalamic nuclei.
Collapse
|
15
|
Zhu H, Yu JJ. Gene expression patterns in the histopathological classification of epithelial ovarian cancer. Exp Ther Med 2010; 1:187-192. [PMID: 23136613 DOI: 10.3892/etm_00000030] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Accepted: 10/05/2009] [Indexed: 01/06/2023] Open
Abstract
The purpose of this study was to screen cancer-related genes and to identify histopathological gene expression patterns as potential biomarkers in human epithelial ovarian cancer (EOC). Fifty genes were screened by reverse-transcription polymerase chain reaction assay with cDNA from 83 EOC tissues and 48 normal ovarian specimens of ovarian cancer patients and evaluated by gel electrophoresis analysis. Twenty expressed genes were assessed by real-time relative-quantity (RQ)-PCR in 30 EOC specimens for gene signature study. Four genes, TAL2, EGF, ILF3 and UBE2I, were investigated for gene expression patterns in histopathological classification of EOC. RQ-value (Ct, ΔCt, ΔΔCt, RQ and gene expression plots) was generated by ABI 7500 Fast System SDS Software (version 1.4). SPSS 15.0 software was used for statistical analysis. Using real-time RQ-PCR, we found that TAL2, EGF, ILF3 and UBE2I demonstrated distinct expression patterns in histological types of epithelial ovarian cancer. The expression of ILF3 and UBE2I in tumors was significantly higher than in normal tissue, with extremely high expression in serous carcinomas compared to mucinous, endometrium and clear cell carcinomas. In addition, ILF3 and UBE2I were overexpressed in advanced stage and advanced grade ovarian cancer, compared to early stage or well-differentiated ovarian cancer. This is the first report of TAL2 and ILF3 expression in the normal human ovary and epithelial ovarian cancer. Our results indicate that overexpression of ILF3 and UBE2I in advanced stage and advanced grade suggest that these two genes may play an important role in tumorigenesis/tumor progression and pathological differentiation of the disease. Notably, ILF3 plays a role in DNA binding activity and transcriptional and post-transcriptional regulation; UBE2I is required in ubiquitination and sumoylation and is involved in DNA repair and apoptosis of cells. Further investigations to reveal the molecular mechanisms related to the activation of ILF3 and UBE2I in the development of EOC are warranted.
Collapse
Affiliation(s)
- Honglan Zhu
- Department of Biochemistry, School of Medicine, Department of Basic Pharmaceutical Sciences, School of Pharmacy and Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA ; ; Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, P.R. China
| | | |
Collapse
|
16
|
Ferran J, de Oliveira ED, Merchán P, Sandoval J, Sánchez-Arrones L, Martínez-De-La-Torre M, Puelles L. Genoarchitectonic profile of developing nuclear groups in the chicken pretectum. J Comp Neurol 2009; 517:405-51. [DOI: 10.1002/cne.22115] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
17
|
Kataoka A, Shimogori T. Fgf8 controls regional identity in the developing thalamus. Development 2008; 135:2873-81. [DOI: 10.1242/dev.021618] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The vertebrate thalamus contains multiple sensory nuclei and serves as a relay station to receive sensory information and project to corresponding cortical areas. During development, the progenitor region of the diencephalon is divided into three parts, p1, p2 (presumptive thalamus) and p3, along its longitudinal axis. Besides the local expression of signaling molecules such as sonic hedgehog (Shh), Wnt proteins and Fgf8, the patterning mechanisms of the thalamic nuclei are largely unknown. Using mouse in utero electroporation to overexpress or inhibit endogenous Fgf8 at the diencephalic p2/p3 border, we revealed that it affected gene expression only in the p2 region without altering overall diencephalic size or the expression of other signaling molecules. We demonstrated that two distinctive populations in p2,which can be distinguished by Ngn2 and Mash1 in early embryonic diencephalon, are controlled by Fgf8 activity in complementary manner. Furthermore, we found that FGF activity shifts thalamic sensory nuclei on the A/P axis in postnatal brain. Moreover, gene expression analysis demonstrated that FGF signaling shifts prethalamic nuclei in complementary manner to the thalamic shift. These findings suggest conserved roles of FGF signaling in patterning along the A/P axis in CNS, and reveal mechanisms of nucleogenesis in the developing thalamus.
Collapse
Affiliation(s)
- Ayane Kataoka
- RIKEN Brain Science Institute, 2-1 Hirosawa, Wako-Shi, Saitama 351-0198,Japan
| | - Tomomi Shimogori
- RIKEN Brain Science Institute, 2-1 Hirosawa, Wako-Shi, Saitama 351-0198,Japan
| |
Collapse
|
18
|
Rabert D, Xiao Y, Yiangou Y, Kreder D, Sangameswaran L, Segal MR, Hunt CA, Birch R, Anand P. Plasticity of gene expression in injured human dorsal root ganglia revealed by GeneChip oligonucleotide microarrays. J Clin Neurosci 2007; 11:289-99. [PMID: 14975420 DOI: 10.1016/j.jocn.2003.05.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2003] [Accepted: 05/09/2003] [Indexed: 11/29/2022]
Abstract
Root avulsion from the spinal cord occurs in brachial plexus lesions. It is the practice to repair such injuries by transferring an intact neighbouring nerve to the distal stump of the damaged nerve; avulsed dorsal root ganglia (DRG) are removed to enable nerve transfer. Such avulsed adult human cervical DRG ( [Formula: see text] ) obtained at surgery were compared to controls, for the first time, using GeneChip oligonucleotide arrays. We report 91 genes whose expression levels are clearly altered by the injury. This first study provides a global assessment of the molecular events or "gene switches" as a consequence of DRG injuries, as the tissues represent a wide range of surgical delay, from 1 to 100 days. A number of these genes are novel with respect to sensory ganglia, while others are known to be involved in neurotransmission, trophism, cytokine functions, signal transduction, myelination, transcription regulation, and apoptosis. Cluster analysis showed that genes involved in the same functional groups are largely positioned close to each other. This study represents an important step in identifying new genes and molecular mechanisms in human DRG, with potential therapeutic relevance for nerve repair and relief of chronic neuropathic pain.
Collapse
|
19
|
Li S, Xiang M. Barhl1 is required for maintenance of a large population of neurons in the zonal layer of the superior colliculus. Dev Dyn 2006; 235:2260-5. [PMID: 16752387 PMCID: PMC2570113 DOI: 10.1002/dvdy.20858] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The mammalian superior colliculus of the midbrain is a brainstem center that integrates sensorimotor signals involved in the control of orienting behaviors. Its structure is characterized by seven well-organized cellular and fibrous layers associated with distinct physiological properties. To date, however, little is known about the molecular bases governing the lamination, differentiation, and survival of superior collicular neurons. Barhl1 is a homeodomain transcription factor that has been demonstrated to play an essential role in maintaining inner ear hair cells, cerebellar granule cells, and precerebellar neurons. We show here that Barhl1 exhibits a select expression pattern in the superior colliculus with positive neurons largely restricted to the zonal layer, as visualized by the beta-galactosidase activity expressed from the lacZ reporter knocked in the Barhl1 locus. Targeted disruption of Barhl1 results in the loss of a large population of neurons from the zonal layer of the superior colliculus, as indicated by reduced beta-galactosidase staining and marker gene expression as well as by increased apoptotic cell death. Taken together, these data suggest that Barhl1 is crucially required for the survival but not for the specification of zonal layer neurons in the superior colliculus.
Collapse
Affiliation(s)
| | - Mengqing Xiang
- Corresponding author: Dr. Mengqing Xiang, Center for Advanced Biotechnology and Medicine, 679 Hoes Lane, Piscataway, NJ 08854, Tel: 732-235-4491, Fax: 732-235-4466, E-mail:
| |
Collapse
|
20
|
Bradley CK, Takano EA, Hall MA, Göthert JR, Harvey AR, Begley CG, van Eekelen JAM. The essential haematopoietic transcription factor Scl is also critical for neuronal development. Eur J Neurosci 2006; 23:1677-89. [PMID: 16623824 DOI: 10.1111/j.1460-9568.2006.04712.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Abstract The basic helix-loop-helix (bHLH) transcription factor Scl displays tissue-restricted expression and is critical for the establishment of the haematopoietic system; loss of Scl results in embryonic death due to absolute anaemia. Scl is also expressed in neurons of the mouse diencephalon, mesencephalon and metencephalon; however, its requirement in those sites remains to be determined. Here we report conditional deletion of Scl in neuronal precursor cells using the Cre/LoxP system. Neuronal-Scl deleted mice died prematurely, were growth retarded and exhibited an altered motor phenotype characterized by hyperactivity and circling. Moreover, ablation of Scl in the nervous system affected brain morphology with abnormal neuronal development in brain regions known to express Scl under normal circumstances; there was an almost complete absence of Scl-null neurons in the hindbrain and partial loss of Scl-null neurons in the thalamus and midbrain from early neurogenesis onwards. Our results demonstrate a crucial role for Scl in the development of Scl-expressing neurons, including gamma-aminobutyric acid (GABA)ergic interneurons. Our study represents one of the first demonstrations of functional overlap of a single bHLH protein that regulates neural and haematopoietic cell development. This finding underlines Scl's critical function in cell fate determination of mesodermal as well as neuroectodermal tissues.
Collapse
Affiliation(s)
- Cara K Bradley
- Telethon Institute for Child Health Research and Centre for Child Health Research at the University of Western Australia, Subiaco WA 6008, Australia
| | | | | | | | | | | | | |
Collapse
|
21
|
Jones HC, Chen GF, Yehia BR, Carter BJ, Akins EJ, Wolpin LC. Single and multiple congenic strains for hydrocephalus in the H-Tx rat. Mamm Genome 2005; 16:251-61. [PMID: 15965786 PMCID: PMC2929525 DOI: 10.1007/s00335-004-2390-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2004] [Accepted: 12/06/2004] [Indexed: 11/25/2022]
Abstract
The H-Tx rat has fetal-onset hydrocephalus with a complex mode of inheritance. Previously, quantitative trait locus mapping using a backcross with Fischer F344 rats demonstrated genetic loci significantly linked to hydrocephalus on Chromosomes 10, 11, and 17. Hydrocephalus was preferentially associated with heterozygous alleles on Chrs 10 and 11 and with homozygous alleles on Chr 17. This study aimed to determine the phenotypic contribution of each locus by constructing single and multiple congenic strains. Single congenic rats were constructed using Fischer F344 as the recipient strain and a marker-assisted protocol. The homozygous strains were maintained for eight generations and the brains examined for dilated ventricles indicative for hydrocephalus. No congenic rats had severe (overt) hydrocephalus. A few pups and a significant number of adults had mild disease. The incidence was significantly higher in the C10 and C17 congenic strains than in the nonhydrocephalic F344 strain. Breeding to F344 to make F.H-Tx C10 or C11 rats heterozygous for the hydrocephalus locus failed to produce progeny with severe disease. Both bicongenic and tricongenic rats of different genotype combinations were constructed by crossing congenic rats. None had severe disease but the frequency of mild hydrocephalus in adults was similar to congenic rats and significantly higher than in the F344 strain. Rats with severe hydrocephalus were recovered in low numbers when single congenic or bicongenic rats were crossed with the parental H-Tx strain. It is concluded that the genetic and epigenetic factors contributing to severe hydrocephalus in the H-Tx strain are more complex than originally anticipated.
Collapse
Affiliation(s)
- Hazel C Jones
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, 32610-0267, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Pinheiro P, Gering M, Patient R. The basic helix-loop-helix transcription factor, Tal2, marks the lateral floor plate of the spinal cord in zebrafish. Gene Expr Patterns 2004; 4:85-92. [PMID: 14678833 DOI: 10.1016/s1567-133x(03)00145-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Basic helix-loop-helix (bHLH) transcription factors play key roles in the development of the central nervous system. Here we report the isolation of a zebrafish gene that encodes a homologue of the mammalian bHLH transcription factor, Tal2. In zebrafish embryos, tal2, like its mammalian homologue, is strongly expressed in the diencephalon and the mesencephalon, with the latter expression located in post-mitotic cells of the tectum. However, in addition to this conserved brain expression, we also detect expression in the floor plate of the spinal cord. By the location of this expression relative to other genes expressed in the floor plate and by analysing expression in a selection of midline mutants, we reveal that tal2 is expressed within the lateral floor plate as opposed to the medial floor plate, and also in more dorsal cells which are distinct from motorneurons and depend on either sonic hedgehog signalling or a signal coming from the lateral floor plate. This is to our knowledge the first report of a gene expressed specifically in lateral cells of the floor plate in the spinal cord.
Collapse
Affiliation(s)
- Philip Pinheiro
- Institute of Genetics, Queens Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
| | | | | |
Collapse
|
23
|
Braquart-Varnier C, Danesin C, Clouscard-Martinato C, Agius E, Escalas N, Benazeraf B, Ai X, Emerson C, Cochard P, Soula C. A subtractive approach to characterize genes with regionalized expression in the gliogenic ventral neuroepithelium: identification of chick Sulfatase 1 as a new oligodendrocyte lineage gene. Mol Cell Neurosci 2004; 25:612-28. [PMID: 15080891 DOI: 10.1016/j.mcn.2003.11.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2003] [Revised: 11/13/2003] [Accepted: 11/20/2003] [Indexed: 11/17/2022] Open
Abstract
To address the question of the origin of glial cells and the mechanisms leading to their specification, we have sought to identify novel genes expressed in glial progenitors. We adopted suppression subtractive hybridization (SSH) to establish a chick cDNA library enriched for genes specifically expressed at 6 days of incubation (E6) in the ventral neuroepithelium, a tissue previously shown to contain glial progenitors. Screens were then undertaken to select differentially expressed cDNAs, and out of 82 unique SSH clones, 21 were confirmed to display a regionalized expression along the dorsoventral axis of the E6 ventral neuroepithelium. Among these, we identified a transcript coding for the chick orthologue of Sulf1, a recently identified cell surface sulfatase, as a new, early marker of oligodendrocyte (OL) precursors in the chick embryonic spinal cord. This study provides groundwork for the further identification of genes involved in glial specification.
Collapse
Affiliation(s)
- Christine Braquart-Varnier
- Centre de Biologie du Développement, UMR5547 CNRS/UPS, Université Paul Sabatier, 31062 Toulouse Cedex, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Chen A, Muzzio IA, Malleret G, Bartsch D, Verbitsky M, Pavlidis P, Yonan AL, Vronskaya S, Grody MB, Cepeda I, Gilliam TC, Kandel ER. Inducible enhancement of memory storage and synaptic plasticity in transgenic mice expressing an inhibitor of ATF4 (CREB-2) and C/EBP proteins. Neuron 2003; 39:655-69. [PMID: 12925279 DOI: 10.1016/s0896-6273(03)00501-4] [Citation(s) in RCA: 213] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
To examine the role of C/EBP-related transcription factors in long-term synaptic plasticity and memory storage, we have used the tetracycline-regulated system and expressed in the forebrain of mice a broad dominant-negative inhibitor of C/EBP (EGFP-AZIP), which preferentially interacts with several inhibiting isoforms of C/EBP. EGFP-AZIP also reduces the expression of ATF4, a distant member of the C/EBP family of transcription factors that is homologous to the Aplysia memory suppressor gene ApCREB-2. Consistent with the removal of inhibitory constraints on transcription, we find an increase in the pattern of gene transcripts in the hippocampus of EGFP-AZIP transgenic mice and both a reversibly enhanced hippocampal-based spatial memory and LTP. These results suggest that several proteins within the C/EBP family including ATF4 (CREB-2) act to constrain long-term synaptic changes and memory formation. Relief of this inhibition lowers the threshold for hippocampal-dependent long-term synaptic potentiation and memory storage in mice.
Collapse
Affiliation(s)
- Amy Chen
- Center for Neurobiology and Behavior, Columbia University, College of Physicians and Surgeons, New York, NY 10032, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
van Eekelen JAM, Bradley CK, Göthert JR, Robb L, Elefanty AG, Begley CG, Harvey AR. Expression pattern of the stem cell leukaemia gene in the CNS of the embryonic and adult mouse. Neuroscience 2003; 122:421-36. [PMID: 14614907 DOI: 10.1016/s0306-4522(03)00571-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The basic helix-loop-helix (bHLH) transcription factor stem cell leukaemia (SCL) is a 'master regulator' of haematopoiesis, where SCL is pivotal in cell fate determination and differentiation. SCL has also been detected in CNS, where other members of the bHLH-family have been shown to be indispensable for neuronal development; however, no detailed expression pattern of SCL has so far been described. We have generated a map of SCL expression in the embryonic and adult mouse brain based on histochemical analysis of LacZ reporter gene expression in sequential sections of brain tissue derived from SCL-LacZ knockin mice. The expression of LacZ was confirmed to reflect SCL expression by in situ hybridisation. LacZ expression was found in a range of different diencephalic, mesencephalic and metencephalic brain nuclei in adult CNS. Co-localisation of LacZ with the neuronal marker NeuN indicated expression in post-mitotic neurons in adulthood. LacZ expression by neurons was confirmed in tissue culture analysis. The nature of the pretectal, midbrain and hindbrain regions expressing LacZ suggest that SCL in adult CNS is potentially involved in processing of visual, auditory and pain related information. During embryogenesis, LacZ expression was similarly confined to thalamus, midbrain and hindbrain. LacZ staining was also evident in parts of the intermediate and marginal zone of the aqueduct and ventricular zone of the fourth ventricle at E12.5 and E14. These cells may represent progenitor stages of differentiating neural cells. Given the presence of SCL in both the developing brain and in post-mitotic neurons, it seems likely that the function of SCL in neuronal differentiation may differ from its function in maintaining the differentiated state of the mature neuron.
Collapse
Affiliation(s)
- J A M van Eekelen
- Centre for Child Health Research and WAIMR, University of Western Australia, at the Telethon Institute for Child Health Research, PO Box 855, West Perth WA 6872, Australia.
| | | | | | | | | | | | | |
Collapse
|
26
|
Bramblett DE, Copeland NG, Jenkins NA, Tsai MJ. BHLHB4 is a bHLH transcriptional regulator in pancreas and brain that marks the dimesencephalic boundary. Genomics 2002; 79:402-12. [PMID: 11863370 DOI: 10.1006/geno.2002.6708] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have cloned a basic helix-loop-helix (bHLH) factor gene, Bhlhb4, from a mouse beta-cell line. Fluorescence in situ hybridization (FISH) and genetic mapping place Bhlhb4 at the telomeric end of mouse chromosome 2 (H3-H4), syntenic to human chromosome 20q13. Based on phylogenetic analysis, BHLHB4 belongs to a new subgroup of bHLH factors including at least four previously identified mouse bHLH factors: BHLHB5, MIST1, OLIG1, OLIG2, and OLIG3. In the developing nervous system, Bhlhb4 was found to mark the dimesencephalic boundary, suggesting that Bhlhb4 may have a role in diencephalic regionalization. In the pancreas, Bhlhb4 is expressed in a transient fashion that suggests a role in the pancreatic endocrine cell lineage. Transfection experiments show that BHLHB4 can repress transcriptional activation mediated through the pancreatic beta-cell specific insulin promoter enhancer RIPE3. Together, these data suggest that BHLHB4 may modulate the expression of genes required for the differentiation and/or maintenance of pancreatic and neuronal cell types.
Collapse
Affiliation(s)
- Debra E Bramblett
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | |
Collapse
|